Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Drug Metab Dispos ; 38(3): 459-73, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20016052

RESUMO

The pharmacokinetics and metabolism of anacetrapib (MK-0859), a novel cholesteryl ester transfer protein inhibitor, were examined in rats and rhesus monkeys. Anacetrapib exhibited a low clearance in both species and a moderate oral bioavailability of approximately 38% in rats and approximately 13% in monkeys. The area under the plasma concentration-time curve in both species increased in a less than dose-proportional manner over an oral dose range of 1 to 500 mg/kg. After oral administration of [(14)C]anacetrapib at 10 mg/kg, approximately 80 and 90% of the radioactive dose was recovered over 48 h postdose from rats and monkeys, respectively. The majority of the administered radioactive dose was excreted unchanged in feces in both species. Biliary excretion of radioactivity accounted for approximately 15% and urinary excretion for less than 2% of the dose. Thirteen metabolites, resulting from oxidative and secondary glucuronic acid conjugation, were identified in rat and monkey bile. The main metabolic pathways consisted of O-demethylation (M1) and hydroxylation on the biphenyl moiety (M2) and hydroxylation on the isopropyl side chain (M3); these hydroxylations were followed by O-glucuronidation of these metabolites. A glutathione adduct (M9), an olefin metabolite (M10), and a propionic acid metabolite (M11) also were identified. In addition to parent anacetrapib, M1, M2, and M3 metabolites were detected in rat but not in monkey plasma. Overall, it appears that anacetrapib exhibits a low-to-moderate degree of absorption after oral dosing and majority of the absorbed dose is eliminated via oxidation to a series of hydroxylated metabolites that undergo conjugation with glucuronic acid before excretion into bile.


Assuntos
Anticolesterolemiantes/farmacocinética , Proteínas de Transferência de Ésteres de Colesterol/antagonistas & inibidores , Oxazolidinonas/farmacocinética , Animais , Anticolesterolemiantes/administração & dosagem , Anticolesterolemiantes/análise , Anticolesterolemiantes/química , Bile/química , Biotransformação , Análise Química do Sangue , Avaliação Pré-Clínica de Medicamentos , Fezes/química , Glucuronídeos/análise , Glucuronídeos/sangue , Glucuronídeos/química , Glucuronídeos/urina , Hidroxilação , Absorção Intestinal , Macaca mulatta , Espectroscopia de Ressonância Magnética , Masculino , Estrutura Molecular , Oxazolidinonas/administração & dosagem , Oxazolidinonas/análise , Oxazolidinonas/química , Ratos , Ratos Sprague-Dawley , Especificidade da Espécie , Espectrometria de Massas em Tandem , Urina/química
2.
Drug Metab Dispos ; 38(3): 474-83, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20016053

RESUMO

Anacetrapib is a novel cholesteryl ester transfer protein inhibitor being developed for the treatment of primary hypercholesterolemia and mixed dyslipidemia. The absorption, distribution, metabolism, and excretion of anacetrapib were investigated in an open-label study in which six healthy male subjects received a single oral dose of 150 mg and 165 microCi of [(14)C]anacetrapib. Plasma, urine, and fecal samples were collected at predetermined times for up to 14 days postdose and were analyzed for total radioactivity, the parent compound, and metabolites. The majority of the administered radioactivity (87%) was eliminated by fecal excretion, with negligible amounts present in urine (0.1%). The peak level of radioactivity in plasma (approximately 2 microM equivalents of [(14)C]anacetrapib) was achieved approximately 4 h postdose. The parent compound was the major radioactive component (79-94% of total radioactivity) in both plasma and feces. Three oxidative metabolites, M1, M2, and M3, were detected in plasma and feces and were identified as the O-demethylated species (M1) and two secondary hydroxylated derivatives of M1 (M2 and M3). Each metabolite was detected at low levels, representing

Assuntos
Anticolesterolemiantes/farmacocinética , Proteínas de Transferência de Ésteres de Colesterol/antagonistas & inibidores , Oxazolidinonas/farmacocinética , Adolescente , Adulto , Anticolesterolemiantes/efeitos adversos , Anticolesterolemiantes/metabolismo , Biotransformação , Citocromo P-450 CYP3A/genética , Citocromo P-450 CYP3A/metabolismo , Inibidores do Citocromo P-450 CYP3A , Inibidores das Enzimas do Citocromo P-450 , Sistema Enzimático do Citocromo P-450/genética , Sistema Enzimático do Citocromo P-450/metabolismo , Fezes/química , Humanos , Isoenzimas/antagonistas & inibidores , Isoenzimas/metabolismo , Masculino , Microssomos Hepáticos/enzimologia , Microssomos Hepáticos/metabolismo , Pessoa de Meia-Idade , Estrutura Molecular , Oxazolidinonas/efeitos adversos , Oxazolidinonas/metabolismo , Proteínas Recombinantes/antagonistas & inibidores , Proteínas Recombinantes/metabolismo , Espectrometria de Massas em Tandem , Adulto Jovem
3.
Bioorg Med Chem Lett ; 19(17): 5033-6, 2009 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-19631535

RESUMO

During our effort to design a receptor binding assay to aid in the elucidation of the molecular mechanism of ezetimibe, we prepared a sulfur-35 containing radioligand which exhibits improved potency over the glucuronide conjugate of ezetimibe in both native enterocyte brush border membranes and membranes from cells expressing recombinant NPC1L1. Herein, we describe the different synthetic strategies which were used to obtain this compound as well as its effectiveness in the aforementioned assay.


Assuntos
Anticolesterolemiantes/química , Azetidinas/química , Proteínas de Membrana/antagonistas & inibidores , Animais , Anticolesterolemiantes/síntese química , Anticolesterolemiantes/farmacologia , Azetidinas/síntese química , Azetidinas/farmacologia , Linhagem Celular , Ezetimiba , Glucuronídeos/química , Humanos , Ligantes , Proteínas de Membrana/metabolismo , Proteínas de Membrana Transportadoras , Camundongos , Ligação Proteica , Ratos , Proteínas Recombinantes/antagonistas & inibidores , Proteínas Recombinantes/metabolismo , Radioisótopos de Enxofre/química
4.
Drug Metab Dispos ; 36(2): 469-73, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17998295

RESUMO

Recent clinical reports have suggested that the cyclooxygenase-2 inhibitor, lumiracoxib (Prexige), may cause a rare but serious hepatotoxicity in patients. In view of the close structural resemblance between lumiracoxib and diclofenac, a widely used nonsteroidal anti-inflammatory drug whose use also has been associated with rare cases of liver injury, it is possible that the toxicity of the two agents may share a common mechanism. Because it is believed that chemically reactive metabolites may play a role as mediators of diclofenac-mediated hepatotoxicity, the present in vitro study was carried out to test the hypothesis that lumiracoxib also undergoes metabolic activation when incubated with liver microsomal preparations and hepatocytes from rats and humans. By means of liquid chromatography tandem mass spectrometry and nuclear magnetic resonance spectrometry techniques, two previously unknown N-acetylcysteine (NAC) conjugates were identified, namely, 3'-NAC-4'-hydroxy lumiracoxib (M1) and 4'-hydroxy-6'-NAC-desfluoro lumiracoxib (M2), the structures of which reveal the intermediacy of an electrophilic quinone imine species. Based on the results of studies with immunoinhibitory antibodies, it was demonstrated that the formation of M1 and M2 in human liver microsomes was catalyzed by cytochrome P450 (P450) 2C9. These findings demonstrate that lumiracoxib is subject to P450-mediated bioactivation in both rat and human liver preparations, leading to the formation of a reactive intermediate analogous to species generated during the metabolism of diclofenac.


Assuntos
Inibidores de Ciclo-Oxigenase 2/metabolismo , Diclofenaco/análogos & derivados , Hepatócitos/metabolismo , Microssomos Hepáticos/metabolismo , Acetilcisteína/farmacologia , Animais , Células Cultivadas , Cromatografia Líquida , Diclofenaco/metabolismo , Humanos , Ratos , Espectrometria de Massas em Tandem
5.
Artigo em Inglês | MEDLINE | ID: mdl-15686997

RESUMO

To support pharmacokinetic studies, a selective and sensitive liquid chromatography/tandem mass spectrometry (LC-MS/MS) method has been developed and validated for the simultaneous determination of a novel KDR kinase inhibitor (1) and its active metabolite (2) in human plasma. The method is fully automated using a Packard MultiPROBE II system and a TomTec Quadra 96 liquid handling workstation to perform sample preparation and solid-phase extraction (SPE). Following the extraction on a mixed-mode SPE using Oasis MCX 96-well plate, the analytes were separated on a Aquasil C18 column (50 mm x 2.1 mm, i.d., 3 microm) with a mobile phase consisting of acetonitrile/ammonium acetate buffer (5 mM, pH 5.0) (60/40, v/v). The run time for each injection was 4.5 min with the retention times of approximately 2.0 and 2.7 min for 1 and 2 respectively, at a flow rate of 0.25 mL/min. A tandem mass spectrometric detection was conducted using multiple reaction monitoring (MRM) under the positive ion mode with a turbo ion-spray interface. The linear ranges of the calibration curves were 0.05-400 ng/mL for 1 and 0.1-400 ng/mL for 2 on a PE Sciex API 4000 LC-MS/MS system. The lower limits of quantitation (LLOQ) of the assay were 0.05 and 0.1 ng/mL for 1 and 2 respectively, when 0.4 mL of plasma was processed. Intra-day assay precision (using five standard curves prepared by spiking compounds to five lots of plasma) was less than 4.9% for 1 and less than 9.6% for 2 on each concentration. Assay accuracy was found to be 95.1-104.6% of nominal for 1 standards and 93.5-105.6% for 2 standards. QC samples were stable when kept at room temperature for 4 h, at -70 degrees C for 10 days, and after three freeze-thaw cycles. The extraction recoveries were 80%, 83% and 84% for 1 and 2 and I.S. respectively, and no significant matrix effects were observed. The method was successfully applied to plasma samples from clinical studies after oral administration of compound 1.


Assuntos
Cromatografia Líquida de Alta Pressão/métodos , Peptídeos e Proteínas de Sinalização Intracelular/sangue , Espectrometria de Massas/métodos , Óxidos/metabolismo , Humanos , Padrões de Referência , Reprodutibilidade dos Testes , Sensibilidade e Especificidade
6.
Cancer Chemother Pharmacol ; 67(6): 1313-21, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20734049

RESUMO

PURPOSE: Because glucocorticoids and the neurokinin-1 receptor antagonist aprepitant influence CYP3A4 activity, this study assessed whether aprepitant added to a 5-HT(3) antagonist and glucocorticoid would affect CYP3A4 induction. METHODS: In this double-blind, 2-period crossover study, 12 subjects were randomized to receive a triple regimen (oral aprepitant [A] 125 mg, intravenous ondansetron [O] 32 mg, and oral dexamethasone [D] 12 mg day 1; A 80 mg and D 8 mg days 2-3; D 8 mg day 4) in 1 of 2 periods, and a dual regimen (O 32 mg and D 20 mg day 1; D 8 mg bid days 2-4); the D dose was adjusted to account for known dexamethasone/aprepitant interaction. Oral (2 mg) and intravenous (1 mg) stable isotope ((13)C(5) (15)N(1))-labeled midazolam were simultaneously given as probes on days -1, 6, 8, 15, and 22 of each period. If the a priori 90% confidence interval for the day 6 geometric mean oral midazolam AUC(0-∞) ratio (triple/dual regimen) of fold-change from baseline was above 0.5, it would be concluded that there was no clinically meaningful between-regimen difference in CYP3A4 activity. RESULTS: Day 6 oral midazolam AUC(0-∞) geometric mean fold-change from baseline was 0.84 (0.30-1.58 with A, 0.46-1.69 without A). The ratio of geometric mean oral midazolam AUC(0-∞) fold-changes was 1.00 (90% confidence interval 0.80, 1.25). CONCLUSIONS: Aprepitant plus a 5-HT(3) antagonist and dexamethasone is unlikely to have a significant additional inductive effect on CYP3A4 activity beyond that of the dual regimen.


Assuntos
Antieméticos/farmacologia , Citocromo P-450 CYP3A/metabolismo , Dexametasona/farmacologia , Morfolinas/farmacologia , Ondansetron/farmacologia , Adulto , Aprepitanto , Estudos Cross-Over , Método Duplo-Cego , Interações Medicamentosas , Feminino , Humanos , Masculino , Midazolam/farmacologia
7.
Pharm Res ; 26(2): 459-68, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19082692

RESUMO

PURPOSE: Gaboxadol, a selective extrasynaptic agonist of the delta-containing gamma-aminobutyric acid type A (GABAA) receptor, is excreted in humans into the urine as parent drug and glucuronide conjugate. The goal of this study was to identify the UDP-Glucuronosyltransferase (UGT) enzymes and the transporters involved in the metabolism and active renal secretion of gaboxadol and its metabolite in humans.Methods. The structure of the glucuronide conjugate of gaboxadol in human urine was identified by LC/MS/MS. Human recombinant UGT isoforms were used to identify the enzymes responsible for the glucuronidation of gaboxadol. Transport of gaboxadol and its glucuronide was evaluated using cell lines and membrane vesicles expressing human organic anion transporters hOAT1 and hOAT3, organic cation transporter hOCT2, and the multidrug resistance proteins MRP2 and MRP4.Results. Our study indicated that the gaboxadol-O-glucuronide was the major metabolite excreted in human urine. UGT1A9, and to a lesser extent UGT1A6, UGT1A7 and UGT1A8, catalyzed the O-glucuronidation of gaboxadol in vitro. Gaboxadol was transported by hOAT1, but not by hOCT2, hOAT3, MRP2, and MRP4. Gaboxadol-O-glucuronide was transported by MRP4, but not MRP2.Conlusion. Gaboxadol could be taken up into the kidney by hOAT1 followed by glucuronidation and efflux of the conjugate into urine via MRP4.


Assuntos
Agonistas GABAérgicos/farmacocinética , Glucuronídeos/metabolismo , Glucuronosiltransferase/metabolismo , Isoxazóis/farmacocinética , Rim/enzimologia , Fígado/enzimologia , Proteínas de Membrana Transportadoras/metabolismo , Administração Oral , Animais , Biotransformação , Células CHO , Cromatografia Líquida , Cricetinae , Cricetulus , Agonistas GABAérgicos/administração & dosagem , Agonistas GABAérgicos/urina , Glucuronosiltransferase/genética , Humanos , Isoenzimas , Isoxazóis/administração & dosagem , Isoxazóis/urina , Proteínas de Membrana Transportadoras/genética , Microssomos Hepáticos/enzimologia , Proteínas Associadas à Resistência a Múltiplos Medicamentos/metabolismo , Proteína 1 Transportadora de Ânions Orgânicos/metabolismo , Proteínas Recombinantes/metabolismo , Espectrometria de Massas em Tandem , Transfecção , UDP-Glucuronosiltransferase 1A
8.
J Pharmacol Exp Ther ; 321(2): 673-83, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-17314201

RESUMO

Sitagliptin, a selective dipeptidyl peptidase 4 inhibitor recently approved for the treatment of type 2 diabetes, is excreted into the urine via active tubular secretion and glomerular filtration in humans. In this report, we demonstrate that sitagliptin is transported by human organic anion transporter hOAT3 (Km=162 microM), organic anion transporting polypeptide OATP4C1, and multidrug resistance (MDR) P-glycoprotein (Pgp), but not by human organic cation transporter 2 hOCT2, hOAT1, oligopeptide transporter hPEPT1, OATP2B1, and the multidrug resistance proteins MRP2 and MRP4. Our studies suggested that hOAT3, OATP4C1, and MDR1 Pgp might play a role in transporting sitagliptin into and out of renal proximal tubule cells, respectively. Sitagliptin did not inhibit hOAT1-mediated cidofovir uptake, but it showed weak inhibition of hOAT3-mediated cimetidine uptake (IC50=160 microM). hOAT3-mediated sitagliptin uptake was inhibited by probenecid, ibuprofen, furosemide, fenofibric acid, quinapril, indapamide, and cimetidine with IC50 values of 5.6, 3.7, 1.7, 2.2, 6.2, 11, and 79 microM, respectively. Sitagliptin did not inhibit Pgp-mediated transport of digoxin, verapamil, ritonavir, quinidine, and vinblastine. Cyclosporine A significantly inhibited Pgp-mediated transport of sitagliptin (IC50=1 microM). Our data indicate that sitagliptin is unlikely to be a perpetrator of drug-drug interactions with Pgp, hOAT1, or hOAT3 substrates at clinically relevant concentrations. Renal secretion of sitagliptin could be inhibited if coadministered with OAT3 inhibitors such as probenecid. However, the magnitude of interactions should be low, and the effects may not be clinically meaningful, due to the high safety margin of sitagliptin.


Assuntos
Membro 1 da Subfamília B de Cassetes de Ligação de ATP/fisiologia , Inibidores de Adenosina Desaminase , Inibidores da Dipeptidil Peptidase IV , Inibidores Enzimáticos/metabolismo , Glicoproteínas/antagonistas & inibidores , Transportadores de Ânions Orgânicos Sódio-Independentes/fisiologia , Transportadores de Ânions Orgânicos/fisiologia , Pirazinas/metabolismo , Triazóis/metabolismo , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/antagonistas & inibidores , Animais , Transporte Biológico , Células CHO , Cricetinae , Cricetulus , Dipeptidil Peptidase 4 , Humanos , Masculino , Proteínas de Membrana Transportadoras/fisiologia , Camundongos , Proteína 2 Associada à Farmacorresistência Múltipla , Proteínas Associadas à Resistência a Múltiplos Medicamentos/fisiologia , Fosfato de Sitagliptina
10.
Drug Metab Dispos ; 34(1): 145-51, 2006 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16251255

RESUMO

Although zomepirac (ZP) and tolmetin (TM) induce anaphylactic reactions and form reactive acyl glucuronides, a direct link between the two events remains obscure. We report herein that, in addition to acyl glucuronidation, both drugs are subject to oxidative bioactivation. Following incubations of ZP with human liver microsomes fortified with NADPH and glutathione (GSH), a metabolite with an MH+ ion at m/z 597 was detected by LC/MS/MS. On the basis of collision-induced dissociation and NMR evidence, the structure of this metabolite was determined to be 5-[4'-chlorobenzoyl]-1,4-dimethyl-3-glutathionylpyrrole-2-acetic acid (ZP-SG), suggesting that the pyrrole moiety of ZP had undergone oxidation to an epoxide intermediate, followed by addition of GSH and loss of the elements of H2O to yield the observed conjugate. The oxidative bioactivation of ZP most likely is catalyzed by cytochrome P450 (P450) 3A4, since the formation of ZP-SG was reduced to approximately 10% of control values following pretreatment of human liver microsomes with ketoconazole or with an inhibitory anti-P450 3A4 IgG. A similar GSH adduct, namely 5-[4'-methylbenzoyl]-1-methyl-3-glutathionylpyrrole-2-acetic acid (TM-SG), was identified when TM was incubated with human liver microsomal preparations. The relevance of these in vitro findings to the in vivo situation was established through the detection of the same thiol adducts in rats treated with ZP and TM, respectively. Taken together, these data suggest that, in addition to the formation of acyl glucuronides, oxidative metabolism of ZP and TM affords reactive species that may haptenize proteins and thereby contribute to the drug-mediated anaphylactic reactions.


Assuntos
Glutationa/metabolismo , Microssomos Hepáticos/metabolismo , Tolmetino/análogos & derivados , Tolmetino/metabolismo , Animais , Cromatografia Líquida/métodos , Feminino , Glutationa/química , Glutationa/farmacologia , Hepatócitos/química , Hepatócitos/efeitos dos fármacos , Hepatócitos/metabolismo , Humanos , Masculino , Microssomos Hepáticos/química , Microssomos Hepáticos/efeitos dos fármacos , NADP/metabolismo , NADP/farmacologia , Oxirredução/efeitos dos fármacos , Ratos , Espectrometria de Massas por Ionização por Electrospray/métodos , Trítio , Troleandomicina/metabolismo , Troleandomicina/farmacologia
11.
Bioorg Med Chem Lett ; 16(4): 872-5, 2006 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-16303300

RESUMO

(3-tert-Butyl-7-(5-methylisoxazol-3-yl)-2-(1-methyl-1H-1,2,4-triazol-5-ylmethoxy)pyrazolo[1,5-d][1,2,4]triazine (1) was recently identified as a functionally selective, inverse agonist at the benzodiazepine site of GABA(A) alpha5 receptors and enhances performance in animal models of cognition. The routes of metabolism of this compound in vivo in rat have been well characterised, the identities of the major metabolites are confirmed by synthesis and their biological profiles were evaluated. An unusual oxidation of the pyrazolo[1,5-d][1,2,4]triazine core to the corresponding pyrazolo[1,5-d][1,2,4]triazin-4(5H)-one scaffold by aldehyde oxidase has been observed.


Assuntos
Agonistas GABAérgicos/metabolismo , Agonistas GABAérgicos/farmacocinética , Agonistas de Receptores de GABA-A , Isoxazóis/metabolismo , Isoxazóis/farmacocinética , Subunidades Proteicas/agonistas , Triazinas/metabolismo , Triazinas/farmacocinética , Animais , Cães , Relação Dose-Resposta a Droga , Agonistas GABAérgicos/síntese química , Hidrólise , Isoxazóis/síntese química , Macaca mulatta , Camundongos , Conformação Molecular , Ratos , Estereoisomerismo , Relação Estrutura-Atividade , Distribuição Tecidual , Triazinas/síntese química
12.
Proc Natl Acad Sci U S A ; 102(23): 8132-7, 2005 Jun 07.
Artigo em Inglês | MEDLINE | ID: mdl-15928087

RESUMO

Ezetimibe is a potent inhibitor of cholesterol absorption that has been approved for the treatment of hypercholesterolemia, but its molecular target has been elusive. Using a genetic approach, we recently identified Niemann-Pick C1-Like 1 (NPC1L1) as a critical mediator of cholesterol absorption and an essential component of the ezetimibe-sensitive pathway. To determine whether NPC1L1 is the direct molecular target of ezetimibe, we have developed a binding assay and shown that labeled ezetimibe glucuronide binds specifically to a single site in brush border membranes and to human embryonic kidney 293 cells expressing NPC1L1. Moreover, the binding affinities of ezetimibe and several key analogs to recombinant NPC1L1 are virtually identical to those observed for native enterocyte membranes. KD values of ezetimibe glucuronide for mouse, rat, rhesus monkey, and human NPC1L1 are 12,000, 540, 40, and 220 nM, respectively. Last, ezetimibe no longer binds to membranes from NPC1L1 knockout mice. These results unequivocally establish NPC1L1 as the direct target of ezetimibe and should facilitate efforts to identify the molecular mechanism of cholesterol transport.


Assuntos
Azetidinas/farmacologia , Proteínas de Membrana/metabolismo , Proteínas de Membrana Transportadoras/metabolismo , Proteínas/metabolismo , Animais , Azetidinas/química , Sítios de Ligação , Linhagem Celular , Membrana Celular/metabolismo , Enterócitos/citologia , Enterócitos/metabolismo , Ezetimiba , Humanos , Mucosa Intestinal/metabolismo , Intestinos/citologia , Macaca mulatta , Proteínas de Membrana/genética , Proteínas de Membrana Transportadoras/deficiência , Proteínas de Membrana Transportadoras/genética , Camundongos , Camundongos Endogâmicos C57BL , Microvilosidades/metabolismo , Doenças de Niemann-Pick , Ligação Proteica , Proteínas/genética , Ratos , Ratos Sprague-Dawley , Especificidade da Espécie
13.
J Pharmacol Exp Ther ; 309(1): 156-64, 2004 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-14722317

RESUMO

Ethinylestradiol (EE) is one of the key constituents of oral contraceptives. Major metabolites of EE in humans are the glucuronide and sulfate conjugates, EE-3-O-glucuronide (EE-G) and EE-3-O-sulfate (EE-S). In the present study, transport of EE-G and EE-S by the human multidrug resistance proteins MRP1, MRP2, and MRP3 was investigated using inside-out membrane vesicles, isolated from Sf9 cells expressing human MRP1, MRP2, or MRP3. Vesicular uptake studies showed that EE-G was not a substrate for MRP1, whereas an ATP-dependent and saturable transport of [(3)H]EE-G was observed in MRP2 (K(m) of 35.1 +/- 3.5 microM) and MRP3 (K(m) of 9.2 +/- 2.3 microM) containing vesicles. EE-S was not transported by either MRP1, MRP2, or MRP3. However, low concentrations of EE-S stimulated MRP2-mediated uptake of ethacrynic acid glutathione. EE-S also stimulated MRP2 and MRP3-mediated uptake of 17beta-estradiol-17beta-D-glucuronide. Interestingly, EE-S stimulated strongly MRP2- and MRP3-mediated uptake of EE-G by increasing its apparent transport affinity, whereas no reciprocal stimulation of EE-S uptake by EE-G was observed. These data indicate that EE-S allosterically stimulates MRP2- and MRP3-mediated transport of EE-G and is not cotransported with EE-G. Our studies demonstrate specific active transport of a pharmacologically relevant drug conjugate by human MRP2 and MRP3, involving complex interactions with other organic anions. We also suggest that caution needs to be taken when using only competition studies as screening tools to identify substrates or inhibitors of MRP-mediated transport.


Assuntos
Estradiol/análogos & derivados , Etinilestradiol/análogos & derivados , Etinilestradiol/farmacocinética , Proteínas de Membrana Transportadoras/metabolismo , Proteínas Associadas à Resistência a Múltiplos Medicamentos/metabolismo , Animais , Transporte Biológico , Linhagem Celular , Membrana Celular/metabolismo , Interações Medicamentosas , Estradiol/farmacologia , Insetos/citologia , Proteína 2 Associada à Farmacorresistência Múltipla , Trítio
14.
Drug Metab Dispos ; 32(8): 848-61, 2004 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15258111

RESUMO

Compound A (3-[2-oxo-3-[3-(5,6,7,8-tetrahydro-[1,8]naphthyrindin-2-yl)propyl]-imidazolidin-1-yl]-3(S)-(6-methoxy-pyridin-3-yl)-propionic acid), a potent and selective antagonist of integrin alpha(v)beta(3) receptor, is under development for treatment of osteoporosis. This study describes metabolism and excretion of A in vivo in rats, dogs, and monkeys, and metabolism of A in vitro in primary hepatocytes from rats, dogs, monkeys, and humans. In all three animal species studied, A was primarily excreted as unchanged drug and, to a lesser degree, as phase I and phase II metabolites. Major biotransformation pathways of A included glucuronidation/glucosylation on the carboxylic group to form acyl-linked glucuronides/glucosides; and oxidation on the tetrahydronaphthyridine moiety to generate a carbinolamine and its further metabolized products. Minor pathways involved O-demethylation and hydroxylations on the alkyl chain. Only in rats, a glutathione adduct of A was also observed, and its formation is proposed to be via an iminium intermediate on the tetrahydronaphthyridine ring. Similar metabolic pathways were observed in the incubates of hepatocytes from the corresponding animals as well as from humans. CYP 3A and 2D subfamilies were capable of metabolizing A to its oxidative products. Overall, these in vitro and in vivo findings should provide useful insight on possible biotransformation pathways of A in humans.


Assuntos
Integrina alfaVbeta3/antagonistas & inibidores , Integrina alfaVbeta3/metabolismo , Animais , Cães , Feminino , Integrina alfaVbeta3/análise , Macaca mulatta , Masculino , Ratos , Ratos Sprague-Dawley
15.
J Pharmacol Exp Ther ; 303(3): 969-78, 2002 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-12438516

RESUMO

Diclofenac is eliminated predominantly (approximately 50%) as its 4'-hydroxylated metabolite in humans, whereas the acyl glucuronide (AG) pathway appears more important in rats (approximately 50%) and dogs (>80-90%). However, previous studies of diclofenac oxidative metabolism in human liver microsomes (HLMs) have yielded pronounced underprediction of human in vivo clearance. We determined the relative quantitative importance of 4'-hydroxy and AG pathways of diclofenac metabolism in rat, dog, and human liver microsomes. Microsomal intrinsic clearance values (CL(int) = V(max)/K(m)) were determined and used to extrapolate the in vivo blood clearance of diclofenac in these species. Clearance of diclofenac was accurately predicted from microsomal data only when both the AG and the 4'-hydroxy pathways were considered. However, the fact that the AG pathway in HLMs accounted for ~75% of the estimated hepatic CL(int) of diclofenac is apparently inconsistent with the 4'-hydroxy diclofenac excretion data in humans. Interestingly, upon incubation with HLMs, significant oxidative metabolism of diclofenac AG, directly to 4'-hydroxy diclofenac AG, was observed. The estimated hepatic CL(int) of this pathway suggested that a significant fraction of the intrahepatically formed diclofenac AG may be converted to its 4'-hydroxy derivative in vivo. Further experiments indicated that this novel oxidative reaction was catalyzed by CYP2C8, as opposed to CYP2C9-catalyzed 4'-hydroxylation of diclofenac. These findings may have general implications in the use of total (free + conjugated) oxidative metabolite excretion for determining primary routes of drug clearance and may question the utility of diclofenac as a probe for phenotyping human CYP2C9 activity in vivo via measurement of its pharmacokinetics and total 4'-hydroxy diclofenac urinary excretion.


Assuntos
Diclofenaco/metabolismo , Glucuronídeos/metabolismo , Microssomos Hepáticos/metabolismo , Animais , Diclofenaco/química , Cães , Glucuronídeos/química , Humanos , Masculino , Fosforilação Oxidativa , Ratos , Ratos Sprague-Dawley
16.
Drug Metab Dispos ; 31(11): 1437-47, 2003 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-14570777

RESUMO

Thrombin is a serine protease that plays a key role in the blood coagulation cascade. Compound I [2-[6-chloro-3-[(2,2-difluoro-2-pyridin-2-ylethyl)amino]-2-oxopyrazin-1(2H)-yl]-N-[(3-fluoropyridin-2-yl)methyl]acetamide] is a potent, selective, and orally bioavailable thrombin inhibitor that is being studied as a possible anticoagulant. Biotransformation studies in rats revealed that 84% of an i.v. dose of I was excreted in the form of two metabolites. Both metabolites were formed by metabolic activation of the pyrazinone ring in I and subsequent rearrangement leading to two novel dihydro-imidazole and imidazolidine derivatives. The structures of these metabolites and their mechanism of formation were elucidated by additional use of two 13C single labels in the pyrazinone ring of I in combination with mass spectrometry and NMR techniques. The metabolite structures described here illustrate the rich metabolic chemistry of the amino-pyrazinone heterocycle.


Assuntos
Fibrinolíticos/metabolismo , Imidazóis/metabolismo , Pirazinas/metabolismo , Animais , Bile/química , Bile/metabolismo , Biotransformação , Isótopos de Carbono/metabolismo , Fibrinolíticos/análise , Fibrinolíticos/química , Imidazóis/análise , Imidazóis/química , Espectroscopia de Ressonância Magnética/métodos , Masculino , Pirazinas/análise , Pirazinas/química , Ratos , Ratos Sprague-Dawley , Espectrometria de Massa de Íon Secundário/métodos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA